Abstract

Abstract Rhabdomyosarcoma (RMS) is a pediatric sarcoma of muscle. RAS pathway activation is the dominant oncogenic driver event in fusion negative RMS, which includes the Embryonal RMS (ERMS). We have previously shown in a transgenic zebrafish model of kRASG12D induced ERMS that tumor-propagating potential is confined to molecularly defined cells that express Myf5, m-Cadherin, c-Met and additional satellite cell markers. MYF5 and MYOD1 are bHLH myogenic regulatory factors (MRFs) that orchestrate skeletal muscle differentiation during development and regeneration. Both are sufficient to reprogram human mesenchymal cells into a myogenic fate. Importantly, these same factors are highly expressed a subset of mouse and human ERMS. Given that Myf5 is highly and specifically expressed in the TPC compartment in our ERMS model, we hypothesized that Myf5 and its transcriptional targets may regulate self-renewal and growth of TPCs. To address this question, we utilized transgenic zebrafish and expressed Myf5 in differentiated ERMS cells that lack proliferative capacity and can not make tumors when transplanted into recipient fish. Induced expression of Myf5 was sufficient to confer tumor-propagating ability to differentiated populations of ERMS cells. These “Induced TPCs” proliferate and generate aggressive tumors that re-express satellite cell markers and yet retain expression of mature muscle markers. Next, we assessed if Myf5 is required for ERMS initiation in the zebrafish model. Remarkably, kRASSG12D-induced ERMS could be generated in Myf5 loss-of-function zebrafish. Moreover, Myf5-deficient ERMS could regrow following transplantation into rag2E450fs immune-compromised zebrafish; however, these tumors are histologically and molecularly distinct when compared with ERMS arising in wild-type fish. Given the redundancy of function between Myf5 and MyoD in muscle and their differential expression in satellite cells and muscle progenitors, our current hypothesis is that Myf5-deficient zebrafish ERMS likely recapitulate a molecularly-distinct class of human ERMS. This idea is consistent with the fact that MYF5 is expressed in only 50% of primary human ERMS and a small fraction of human cell lines. Importantly, knock down experiments in human ERMS cell lines confirm independent roles of either MYF5 or MYOD1 in maintenance of ERMS cell viability in vitro. Collectively, our results support a previously unappreciated role for bHLH MRFs in ERMS cell survival and intra-tumor functional heterogeneity and suggest that myogenic factor expression may define unique subtypes of ERMS. Citation Format: Ines M. Tenente, Myron Ignatius, Eleanor Chen, Madeline Hayes, David M. Langenau. Myogenic regulatory factors and their role in embryonal rhabdomyosarcoma. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 5153. doi:10.1158/1538-7445.AM2015-5153

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call